Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Nano ; 17(14): 13594-13610, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37458484

ABSTRACT

Delivery of mRNA-based therapeutics to the perinatal brain holds great potential in treating congenital brain diseases. However, nonviral delivery platforms that facilitate nucleic acid delivery in this environment have yet to be rigorously studied. Here, we screen a diverse library of ionizable lipid nanoparticles (LNPs) via intracerebroventricular (ICV) injection in both fetal and neonatal mice and identify an LNP formulation with greater functional mRNA delivery in the perinatal brain than an FDA-approved industry standard LNP. Following in vitro optimization of the top-performing LNP (C3 LNP) for codelivery of an adenine base editing platform, we improve the biochemical phenotype of a lysosomal storage disease in the neonatal mouse brain, exhibit proof-of-principle mRNA brain transfection in vivo in a fetal nonhuman primate model, and demonstrate the translational potential of C3 LNPs ex vivo in human patient-derived brain tissues. These LNPs may provide a clinically translatable platform for in utero and postnatal mRNA therapies including gene editing in the brain.


Subject(s)
Brain Diseases , Nanoparticles , Mice , Humans , Animals , Gene Editing , Lipids , Liposomes , RNA, Messenger/genetics , RNA, Small Interfering/genetics
2.
J Exp Med ; 220(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36584406

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) can replace endogenous microglia with circulation-derived macrophages but has high mortality. To mitigate the risks of HSCT and expand the potential for microglia replacement, we engineered an inhibitor-resistant CSF1R that enables robust microglia replacement. A glycine to alanine substitution at position 795 of human CSF1R (G795A) confers resistance to multiple CSF1R inhibitors, including PLX3397 and PLX5622. Biochemical and cell-based assays show no discernable gain or loss of function. G795A- but not wildtype-CSF1R expressing macrophages efficiently engraft the brain of PLX3397-treated mice and persist after cessation of inhibitor treatment. To gauge translational potential, we CRISPR engineered human-induced pluripotent stem cell-derived microglia (iMG) to express G795A. Xenotransplantation studies demonstrate that G795A-iMG exhibit nearly identical gene expression to wildtype iMG, respond to inflammatory stimuli, and progressively expand in the presence of PLX3397, replacing endogenous microglia to fully occupy the brain. In sum, we engineered a human CSF1R variant that enables nontoxic, cell type, and tissue-specific replacement of microglia.


Subject(s)
Microglia , Protein Engineering , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Humans , Mice , Aminopyridines/pharmacology , Brain/metabolism , Microglia/metabolism , Protein Engineering/methods , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Cell- and Tissue-Based Therapy/methods
3.
Neuron ; 98(6): 1170-1183.e8, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29861285

ABSTRACT

Microglia, the brain's resident macrophages, are dynamic CNS custodians with surprising origins in the extra-embryonic yolk sac. The consequences of their distinct ontogeny are unknown but critical to understanding and treating brain diseases. We created a brain macrophage transplantation system to disentangle how environment and ontogeny specify microglial identity. We find that donor cells extensively engraft in the CNS of microglia-deficient mice, and even after exposure to a cell culture environment, microglia fully regain their identity when returned to the CNS. Though transplanted macrophages from multiple tissues can express microglial genes in the brain, only those of yolk-sac origin fully attain microglial identity. Transplanted macrophages of inappropriate origin, including primary human cells in a humanized host, express disease-associated genes and specific ontogeny markers. Through brain macrophage transplantation, we discover new principles of microglial identity that have broad applications to the study of disease and development of myeloid cell therapies.


Subject(s)
Brain/cytology , Cell Lineage , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Microglia/cytology , Animals , Brain/metabolism , Central Nervous System , Humans , Macrophages/metabolism , Mice , Mice, Knockout , Microglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor
4.
Curr Opin Virol ; 16: 15-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26590692

ABSTRACT

Natural killer (NK) cells are important in immune defense against virus infections. This is predominantly considered a function of rapid, innate NK-cell killing of virus-infected cells. However, NK cells also prime other immune cells through the release of interferon gamma (IFN-γ) and other cytokines. Additionally, NK cells share features with long-lived adaptive immune cells and can impact disease pathogenesis through the inhibition of adaptive immune responses by virus-specific T and B cells. The relative contributions of these diverse and conflicting functions of NK cells in humans are poorly defined and likely context-dependent, thereby complicating the development of therapeutic interventions. Here we focus on the contributions of NK cells to disease in diverse virus infections germane to human health.


Subject(s)
Host-Pathogen Interactions/immunology , Killer Cells, Natural/immunology , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Killer Cells, Natural/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Virus Diseases/metabolism , Viruses/classification
SELECTION OF CITATIONS
SEARCH DETAIL
...